Download as pdf or txt
Download as pdf or txt
You are on page 1of 34

Chapter 16

Gene Delivery Systems: Recent Progress


in Viral and Non-Viral Therapy

Erdal Cevher, Ali Demir Sezer and Emre Şefik Çağlar

Additional information is available at the end of the chapter

http://dx.doi.org/10.5772/53392

1. Introduction
Thanks to the changes in medicine, pharmacological treatment rapidly progresses into new
fields. There is an emphasis on the development of treatment methods to eliminate
underlying factors rather than to treat the symptoms of a disease. Therefore, research is
increasingly utilizing knowledge from the field of genetics. Genetic mutation and deletion
lead to many genetic disorders. Genetic disorders in metabolic pathways, regulation of cell
cycle, ligand/receptor function, cell skeleton and extracellular proteins cause serious
diseases (Yaron et al., 1997).

Both genetic and acquired diseases may be treated using gene therapy. Genetic diseases are
generally caused by deletion or mutation of a single cell. Conversely, in acquired diseases, a
single gene cannot be defined as the sole cause of a disease. Gene therapy has increased in
prominence, and has shown great potential in treating acquired and genetic diseases
(Conwell and Huang, 2005).

Gene delivery systems are categorized as: viral-based, non-viral-based and combined hybrid
systems. Viral-mediated gene delivery systems consist of viruses that are modified to be
replication-deficient, but which can deliver DNA for expression. Adenoviruses, retroviruses,
and lentiviruses are used as viral gene-delivery vectors (Escors and Breckpot, 2010).

Non-viral gene delivery systems were introduced as an alternative to viral-based systems.


One of the most important advantages of these systems is improved transfection. Non-viral
systems are categorized according to preparation, as physical or chemical types. The most
common physical methods are microinjection, electroporation, ultrasound, gene gun, and
hydrodynamic applications. In general terms, physical methods refer to delivery of the gene
via the application of physical force to increase permeability of the cell membrane. In
contrast, chemical methods utilize natural or synthetic carriers to deliver genes into cells. In

© 2012 Cevher et al., licensee InTech. This is an open access chapter distributed under the terms of the
Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits
unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
438 Recent Advances in Novel Drug Carrier Systems

this method, polymers, liposomes, dendrimers, and cationic lipid systems are used as gene
delivery systems (Miyazaki et al., 2006; Prokop and Davidson, 2007).

2. Gene therapy
This chapter provides general information on gene delivery systems, how they are used,
their relative merits, and selection of the most appropriate methods for new studies on gene
therapy.

The drug sector is entering a new era that will enable treatment of the underlying cause
rather than the symptoms of a disease. Many human diseases result from mutations or
deletions, in genes, which lead to disorders in metabolic pathways, ligand/receptor function,
cell cycle regulation, cell skeleton or extra-cellular protein structure and function (Sullivan,
2003). With gene therapy, the disease can be treated by the injection of exogenous nucleic
acid sequences designed to target the diseased tissues of the body (Yaron et al., 1997).

Diseases that can be treated by gene therapy are categorized as either genetic or acquired.
Genetic diseases are those which are typically caused by the mutation or deletion of a single
cell. Conversely, a single gene is not defined as the sole cause of acquired diseases.
Although gene therapy was initially used to treat genetic disorders only, it is now used to
treat a wide range of diseases such as cancer, peripheral vascular diseases, arthritis,
neurodegenerative disorders and AIDS (Mhashilkar et al., 2001). The expression of a single
cell, directly delivered to the cells by a gene delivery system can potentially eliminate a
disease. Prior to gene therapy studies, there was no alternative treatment for genetic
disorders. Today, it is possible to correct genetic mutation with gene therapy studies
(Sullivan, 2003).

2.1. Chronology
The term gene therapy was first introduced at the International Congress of Genetics (Yaron
et al., 1997). The techniques utilized by Gregor Mendel in the 1850s, which were then
developed by Ronald Fischer at the beginning of the twentieth century, formed the turning
points in genetics. The work of both Mendel and Fischer laid the foundations of genealogy.
The material they studied was later termed as gene by the Danish botanist Wilhelm
Johannsen (Yaron et al., 1997). Towards the end of the 1970s, the background of the majority
of genetic disorders was understood, and gene therapy came to the fore. The first gene
therapy trial in humans was conducted at the beginning of the 1970s, and it was observed
that naturally occurring DNA and RNA tumor viruses successfully delivered new genetic
information to the genomes of mammal cells (Escors and Breckpot, 2010).

Due to developments in the science of genetics, at the beginning of the twentieth century, it
was understood that diseases such as hemophilia were genetic diseases. Similarly, it was
found that diseases such as colon cancer, diabetes, and retinoblastoma were also genetic-
based diseases. In the 1980s, gene transfer to mammalian cells came to the fore after the
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 439

development of retroviral vectors (Yaron et al., 1997). In the mid-1980s, gene transfer to
mammalian cells became a routine procedure. Retrovirus-based gene therapies brought
significant advantages, as they can stably integrate their genomes to host-cell chromosomes.
After the 1980s, DNA was defined as a genetic material. Later, it was structurally analyzed
and further advances allowed the modification of genetic code. These discoveries on genetic
material made cloning possible (Escors and Breckpot, 2010).

In 2006, melanoma was successfully treated for the first time. The treatment used a
retroviral formulation of melanoma antigen (MART-1)-specific T-cell receptor (TCR), which
encodes α- and β- chains. In recent years, great advances have been made in gene therapy,
ranging from somatic cloning and completion of human genome project to the discovery of
microRNA-based gene-regulating systems (Escors and Breckpot, 2010).

The use of non-viral gene delivery systems in gene therapy also significantly increased in
recent years. Naked plasmid DNA, coated with gold particles was effectively introduced to
cells using the non-viral gene gun technique. This technique was first used in plant cells in
1987, and is today commonly used in mammal cells and tissues. One of the most
successful current gene therapy techniques is hydrodynamic injection (Willemejane and
Mir, 2009).

2.2. Gene delivery systems


Gene delivery systems use various methods to allow uptake of the gene that has been
selected to target the cell (Conwell and Huang, 2005). The successful design of a gene
delivery system requires complete understanding of the interaction mechanism between the
target cell and delivery system. Understanding intercellular traffic and targeting mechanism
is the most important factor in designing a more effective gene delivery system. Cell
targeting refers to delivery of the therapeutic agent to a specific compartment or organelle of
the cell. It is the most commonly used mechanism in endocytosis gene therapy, particularly
in cellular uptake of non-viral gene delivery systems (Prokop and Davidson, 2007). After the
cellular uptake of the delivery system by endocytosis, cellular release takes place to initiate
DNA transcription and translation, and to produce the related protein. A successful gene
delivery procedure involves minimizing potential inhibitory inflammatory response while
also overcoming certain barriers at each step of the gene delivery procedure, in order to
optimize gene activity (Conwell and Huang, 2005).

Viral gene delivery systems consist of viruses that are modified to be replication-deficient
which were made unable to replicate by redesigning which can deliver the genes to the cells
to provide expression. Adenoviruses, retroviruses, and lentiviruses are used for viral gene
delivery. Viral systems have advantages such as constant expression and expression of
therapeutic genes (Sullivan, 2003). However, there are some limitations that restrict the use
of these systems, which particularly include the use of viruses in production,
immunogenicity, toxicity and lack of optimization in large-scale production (Witlox et al.,
2007) (Figure 1).
440 Recent Advances in Novel Drug Carrier Systems

Figure 1. Gene therapy strategies; mutation compensation, suicide gene therapy and immuno-
potentiation (Witlox et al., 2007).

Non-viral gene delivery systems were developed as an alternative to viral-based systems.


One of the most important advantages of these systems is that they develop transfection.
Non-viral gene delivery systems are divided into two categories: physical and chemical.
Microinjection, electroporation, gene gun, ultrasound-mediated methods, and
hydrodynamic systems are the most widely used physical methods. Physical methods
involve the use of physical force to increase the permeability of the cell membrane and allow
the gene to enter the cell. The primary advantage of physical methods is that they are easy to
use and reliable. However, they also have the disadvantage of causing tissue damage in
some applications.

Chemical methods involve the use of carriers prepared from synthetic or natural
compounds for gene delivery into the cell, including synthetic and natural polymers,
liposomes, dendrimers, synthetic proteins, and cationic lipids. The biggest advantages of
these systems are that they are non-immunogenic and generally have low toxicity.

2.2.1. Viral gene delivery systems


Since a large number of viruses have appropriate mechanisms for transfer of genetic
material to the target cell, current gene technologies concentrated on the use of viral vectors
that provide high transduction effectiveness and advanced level of gene expression. The
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 441

optimal design of a viral vector depends on the types of virus to be used (Wunderbaldinger
et al., 2000).

For safe application of in vivo gene therapy, firstly some problems should be eliminated.
Therapeutic factors should only be expressed in related cell types, and should not cause any
undesired effect in healthy cells (Yaron et al., 1997).

2.2.1.1. Adenoviral systems

Adenoviruses (Ad) were first discovered in 1953 by isolation from human adenoid tissue
cultures (Campos and Barry, 2007; Majhen and Ambriovic-Ristov, 2006). They are
commonly used as gene vectors (Dinh et al., 2005). C group adenoviruses Ad2 and Ad5 are
the most widely studied adenoviruses. The capsid of an adenovirus determines virus
tropism. Groups A and C–F first bind to highly-expressed coxsackie virus B-adenovirus
receptor, and thus realize their high infectivity in many tissues. In contrast, group B binds to
complement-regulatory protein CD46. Adenoviruses are replicated and produce virions,
which contain the nucleus of the infected cell (Dinh et al., 2005). These vectors have the
ability to be replicated, and purification of the vectors generally involves easier and shorter
processes (Armendariz-Borunda et al., 2011).

Adenoviruses are well-characterized, non-integrated, ~26–40 kb in length, relatively large,


non-enveloped, linear dsDNA viruses coated with icosahedral particle, with a diameter of ~
950 Å (excluding elongated fiber proteins) and a molecular weight of approximately
150MDa. They are unreplicated, and infect cells quickly. Adenoviral particles do not contain
lipid or membrane; therefore they are stable in solvents such as ether or ethanol. The use of
adenoviruses, which are one of the first developed vector systems for gene expression, is a
great discovery (Campos and Barry, 2007).

Adenoviruses have important characteristics, which make them indispensable for gene
transfer. The most important ones are their well-known molecular biology; delivery capacity
of foreign DNA fragments up to 36kb; and ability to transfect DNA into many cell types
(Sullivan, 2003).

Adenoviruses are one of the largest and most complex viruses, whose Ad structure was
analyzed with cryo-electron microscopy and X-ray diffractometry. An Ad genome of
approximately 36kb codes more than forty genomes; however, only 12 of these were
demonstrated as the component of the virus particle. As seen in Figure 2 it was reported that
crystal structures of single Ad proteins contained fiber knob, shaft, domains, penton base,
hexon, and cysteine protease. Ad capsid consists of 252 sub-units called capsomeres, which
contain 240 hexonproteins and 12 of the penton base. In addition, the capsid contains pIIIa,
pVI, PVIII, and pIX proteins. Each of the 12 capsid corners contains penton bases wrapped
by 5 hexons. The penton base serves as a hook for the fiber protein, which projects from the
virus like an antenna. Fiber is a homotrimer, where 3 identical polypeptides bind in the
same direction, and which contains 3 structurally and functionally different domains: N-
terminus, which binds the fiber non-covalently to the penton base; C-terminus, which gives
the disc-like knob form, which is responsible for binding to the receptor; and the rod-like
442 Recent Advances in Novel Drug Carrier Systems

shaft, which varies in length according to the serotype of the virus (Medina-Kauwe LK.
2003; Majhen and Ambriovic-Ristov, 2006).

Figure 2. Schematic of the adenovirus capsid. (A) Whole capsid identifying fiber, penton base, and
hexon. (B) Enlargement of circled region in (A), showing homotrimeric fiber bound to homopentameric
penton base. (C) Fiber monomer, identifying tail, shaft, and knob domains. Pictures are not drawn to
scale (Medina-Kauwe LK. 2003).

A viral genome is in a condensed state inside cell nucleus, together with protein V, VII and
X, and the 5′ terminus of the AdDNA is covalently linked to a terminal protein (TP). The
terminal protein (TP) functions as the initiation primer in viral DNA replication. An Ad
genome consists of 5 early (E1A, E1B, E2, E3 and E4), 4 interim (IVa2, IX, VAI, VAII) and one
late transcriptional unit. E1A is the first viral transcriptional unit, which will be expressed
after entering the cell. E1A proteins activate other transearly transcription units, which will
create an optimum medium for virus replication by causing the products to enter in S phase
of the cell E1B proteins bind to p53, Bak, and Bax proteins, and allows the infected cell
survive by inhibiting p53 dependent apoptosis. The E2A unit encodes the proteins that
function in viral genome replication, including DNA polymerase, prethermal protein, and
single-stranded DNA-binding protein. While the E4 transcription unit encodes the proteins
that affect cell cycle control and transformation, E3 protein disrupts the immune response of
the host and ensures the continuation of the infected cell (Majhen and Ambriovic-Ristov,
2006).

The nucleus is surrounded by an icosahedral capsid with a symmetrical structure, formed


by the non-covalent interactions between 7 proteins (II, III, IIIa, IV, VI, VIII, and IX) (Smith
et al., 2010). Adenoviral structural proteins are responsible for the stabilization of genome
and encapsidation of the nucleoprotein nucleus. The icosahedral capsid consists of seven
peptides:trimerichexons, which are found in complex form with three minor capsid
polyproteins (VI, VIII and IX), which provides stabilization (II); pentonbase (III); receptor
binding fiber (IV) and penton-bound protein which serves as a bridge between protein and
the hexon base (III). The fiber consists of 3 domains which are tail in N-terminus, rod-like
shaft and globular knob in C-terminus (Couglan et al., 2010). Adenoviruses enter target cells
via receptor-mediated endocytosis. Instead of combining its own DNA with the genomes of
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 443

the host cell, the adenovirus, remains as an episome within the infected cell. This provides
high efficiency (10); however, it should not be ignored, since it will restrict the longevity of
expressed trans genes; in other words, protein expression will quickly cease (Yaron et al.,
1997; Muzzonigro et al., 1999).

Penton and fiber proteins of virus capsid interact with the coxsackievirus-adenovirus
receptor cell surface protein to provide cell binding. These proteins are then localized in
clathrin-coated pits through NPXY motive of β3 and β5 integrin sub units of the cell.
Dinamine, which is systolic GTPase, mediates cellular uptake of the virus into the cell via
endocytosis by providing construction and development of clathrin-coated pits. Viral capsid
proteins dissociate prior to endocytosis, and the pH value of the viral endosome decreases due
to proton pumps. At pH 6.0, the virus has the ability to detach from the vesicle and enter the
cytosol. Capsid proteins of the virus move towards the nucleus through microtubules and
fragment in the following stages. Although a considerable part of the capsid proteins remain
in the periphery of the nucleus, viral DNA crosses via nuclear pores (Ziello et al., 2010).

For successful delivery of DNA to the nucleus, viruses must facilitate cell-specific binding,
endocytosis internalization, propagation from endocytic vesicles to cytosol, delivery into
cytoplasm, translocation from one terminus of the nuclear envelope to other, and finally
expression of the delivered gene (Dinh et al., 2005).

Direct injection or bolus delivery through inhalation is the easiest form of viral delivery.
However, since the virus will spread from the injection area, a high dose is required to achieve
therapeutic effectiveness. The spread of the virus from the injection site limits the regional
effectiveness and immune response. Adenoviral vectors can be targeted via selective
modification of coat proteins. There is a relatively high level of protein expression following
the transduction, equal to approximately 35% of total cellular protein (Zhang et al., 2009).

Adenoviruses have many natural properties that enable them to be used as a vector for on
colitic, vaccine, or gene therapy. Non-enveloped viruses can be kept in lyophilized form in a
stable state inside a flacon tube or capsule; they can be transported without cold chain; by
mediating in high transduction effectiveness in dividing and non-dividing cells, they can
form 104 virus particles per infected cell (Khare et al., 2011). Adenoviruses achieve suitable
transduction through a high level of expression and become beneficial in in vivo conditions
(Muzzonigro et al., 1999).
Adenoviruses have been one of the most promising methods for high effectiveness in in vivo
gene therapy. For example, following systemic vector delivery, transduction levels of
hepatocytes were observed by adenoviral systems. These are some of the most effective
vectors for gene delivery to various organs; however, they have very significant
disadvantages. Some target cells have low ratios of appropriate primary and/or secondary
adenoviral receptors, and this requires a high dose of vector application to cause target-cell
cytotoxicity. Additionally, non-discriminating tropism of the virus can also lead to
transduction to untargeted cells (Reynolds et al., 1999). The most serious problem in the use
of Ad vectors is their tendency to cause strong immune and inflammatory responses at high
doses (Navarro et al., 2008).
444 Recent Advances in Novel Drug Carrier Systems

2.2.1.2. Retroviral systems

Retroviruses are diploid, single-stranded, circular-enveloped RNA viruses of the family


Retroviridae, with a genome of 7–11 kb, and a diameter of approximately 80–120 nm (Osten
et al., 2007; Escors and Brecpot, 2010). The Retroviridae family is divided into two sub-
families: Orthoretrovirinae and Spumaretrovirinae species (Osten et al., 2007). Retroviruses
cause diseases such as AIDS, leukemia, and cancer; however, their use as a vector in gene
therapy brought new developments in treatment (Pages and Danos, 2003).

The ability of retrovirus-based gene delivery vectors to carry foreign genetic material was
first realized in the early 1980s (El-Aneed, 2004; Escors and Brecpot, 2010). Retroviruses are
viruses that integrate with host genome to produce viral proteins (gag, pol, env) that are
extracted during gene delivery (El-Aneed, 2004). Retroviral vectors have the capacity to
delivery DNA up to 8 kb (Navarro et al., 2008). The most commonly used retroviruses are
the Moloney murine leukemia virus species, which have the capacity to deliver exogenous
genetic material up to approximately 9 kb (Muzzonigro et al., 1999).
A virion nucleus consists of circular or triangular gag-encoded capsid proteins. They are
coated with gag-encoded nucleocapsid protein. The nucleus contains pol-encoded enzymes,
reverse transcriptase, and integrase. Retrovirus genomes have various characteristics. For
example, these viruses are diploid in the real sense, and their genomes are produced by
cellular transcriptional mechanism. In addition, these viruses require cellular (tRNA) for
replication (Luban, 2000). The genome of these (+) sense RNA viruses is not directly
processed as mRNA immediately after infection Considering the structure of the genome,
retroviruses are categorized as either simple or complex forms. Simple and complex
retroviruses encode gag (group-specific antigen), pro (protease), and pol (polymerase)
genes; complex retroviruses also encode a large number of additional genes. Retroviruses
were the first viruses to be modified for gene delivery, and have also been used in the
majority of clinical trials of gene therapy (Hu and Pathak, 2000).

Most of retroviral vectors used in clinical studies are based on the Moloney murine
leukemia virus (MMLV). MMLV is a well-studied and characterized virus; its genome
encodes three polyproteins that are required in trans form for replication and packaging:
gag, pol and env (Escors and Brecpot, 2010).

An ideal retroviral vector for gene delivery should be cell-specific, regulated, and safe.
Effectiveness of delivery is important, as it will also determine the effectiveness of therapy
(Hu and Pathak, 2000). Retroviruses have a lipid envelope. In order to enter a host cell, they
use the interactions between cellular receptors and virally encoded proteins, which are
embedded in the membrane (Hu and Pathak, 2000). Combination of chimeric envelope
protein with viral particles allows the retrovirus to bind receptor-positive target cells (Yi et
al., 2011). SU in the virion surface binds to CD, which is a cell-surface protein expressed by
some T-cells and is found in macrophages. Binding to CD4 induces some changes in the
envelope protein (SU) of the domain of epidermal growth factor (EGF), which allows for
interaction with chemokine receptors (CKR). CKRs are a family of cell-surface-G-proteins
functioning as receptors to stain molecules called chemokines.
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 445

Infusion of viral and cellular membranes starts the internalization of the viral nucleus (Yi et
al., 2011); the viral envelope combines with the cell membrane. Envelope glycoprotein (Env)
of retroviruses is responsible for determining tropism. Binding of Env to cellular receptor
and fusion of viral and cellular membranes are the first stages of infection. Trials that
targeted retroviruses by adding single-stranded antibodies or Env cell-binding ligands
showed limited success. The addition of such large ligands to Env protein prevents Env
from binding to the virion (Bupp and Roth, 2002). Env protein takes a particle-coated
structure and releases the viral nucleus into the cytoplasm. Single-stranded DNA gets
reversely transcripted to double-stranded DNA in the nucleus (Osten et al., 2007; Karlsson et
al., 1987). Viral RNA is reverse transcribed to a DNA copy using virally-encoded reverse-
transcriptase enzyme in the virion (Hu and Pathak, 2000). Retroviruses follow a different
replication cycle through transformation of single-stranded RNAs into double-stranded
DNA in the infected cell. Viral DNA synthesis takes place in cytoplasm by virion-dependent
DNA polymerase, which is termed reverse transcriptase. Viral DNA enters the nucleus,
where takes the ring form for the first time, and serves as a draft for RNA synthesis (Olsen
and Swanstorm, 1985). The first DNA product is a linear double layer molecule, which
forms during DNA synthesis. Within the nucleus, some linear DNA is turned into many
different forms of ring DNA; these mainly contain two copies of LTR units in tandem, or a
single copy of LTR unit (Olsen and Swanstorm, 1985). The end of each LTR unit creates
short, deficient repetitions; this represents the strand domain required for this integration
(Olsen and Swanstorm, 1985), and is delivered to the nucleus. This delivery takes place
through cell division for oncoretroviruses, and through active transport for lentiviruses.
Here, lentiviruses are observed to be more advantageous than oncoretroviruses, as they have
the ability to deliver genes to non-dividing cells. When viral DNA enters the nucleus, it
integrates with the DNA of the host cell (provirus). Retroviruses introduce their genetic
material to the host cell genome in a stabile manner during mitotic division. Since these types
of retroviruses only transfer genes to dividing cells, many procedures that utilize retroviruses
are applied ex vivo. However, more recent lentiviral-based retroviral vectors reduce these
restrictions (Muzzonigro et al., 1999). Most retroviruses infect cells that can be actively divided
during mitotic division. This property protects normal tissue, and although it naturally targets
the tumor, all tumors contain non-dividing cells in G0, which is the resting phase of the tumor
cell cycle (Hu and Pathak, 2000; Kitamura et al., 2003; El-Aneed, 2004) (Figure 3).
Infected cells are then transcribed and spliced. Full-length viral RNA (which is the RNA that
encodes all proteins) is delivered to cytoplasm and translated; other cellular procedures are
modification and delivery of RNA from the nucleus (Hu and Pathak, 2000). Spliced full-
length viral RNA is packaged into viral particles. Retroviruses are RNA viruses that are
replicated over integrated DNA intermediate products. Retroviral particles surround two
copies of full-length viral RNA, containing all of the genetic information required for virus
replication, with capsid (Hu and Pathak, 2000). Virologic and genetic studies indicated that
specific packaging of retroviral genomic RNA was carried out through the interaction with
the nucleocapsid (NC) domain of Gag polyprotein. Retroviral NC domains are generally
very simple, and contain one or two zinc knob motifs consisting of C-C-H-C sequences. Zinc
knobs form metal-coordinated reverse turn, stabilized by NH-S hydrogen bonds. Most
retroviral zinc knobs contain a hydrophobic cut on the surface of the mini-globular domain,
446 Recent Advances in Novel Drug Carrier Systems

Figure 3. Structures of retrovirus vectors. (A) Basic structure of retrovirus vectors and two transcripts
from the vector. In the replication competent MMLV, the Gag-Pol and the Env proteins are translated
from the genomic RNA and the subgenomic RNA, respectively. The subgenomic RNA is a spliced form
RNA, and the splicing occurs from the splice donor site (SD) to the splice acceptor site (SA). Both 5′
and 3′ LTRs consist of U3, R, and U5 regions. Ψ = packaging signal; Δgag = truncated gag sequence.
(B) Structures of versatile pMXs-derived vectors. LTR = long terminal repeat; Ψ = packaging signal;
MCS = multi-cloning site; IRES = internal ribosomal entry site; GFP = green fluorescent protein'
neor = neomycin-resistant gene; puror = puromycin-resistant gene. (C) Structures of pMXs, pMYs, and
pMZs vectors. The 5′ LTR, primer binding site (PBS), and the extended packaging signal of pMXs are
derived from the MFG vector. PBS used in pMYs and pMZs are derived from MESV (murine embryonic
stem cell virus) and binds tRNA-glu instead of tRNA-pro. MCS is designed for cDNA library
construction and is preceded by triple stop codons (not shown). White box = MMLV LTR; gray
box = PCMV LTR; hatched box = MPSV LTR or SFFVp LTR. MCS = multi-cloning site (Kitamura et al.,
2003).
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 447

recognizing the specific structure of RNA or DNA. Base N- and C- terminal tails of
nucleocapsid domains are conformationally indurable (Miyazaki et al., 2011). Retroviral
genome packaging is generally localized between the binding donor (SD) region and the gag
start codon in the 5′ head region. It should be noted that the packaging sign generally
coincides with the dimerization region, which indicates that packaging combines with
genome dimerization (Miyazaki et al., 2011). Inhibition of genome dimerization through
addition or deletion of mutations at dimer initiation sites (DIS) causes a significant decrease
in genome packaging. Studies of mutant viruses containing two retroviral genomes showed
that these were non-covalently dimerized in progen viruses. 5′ untranslated region (UTR)
packages monomeric genome, which indicates that the genome packaging procedure occurs
via the interaction of two 5′ UTR (Miyazaki et al., 2011). Virion maturation occurs by the
budding of the particle from the cell (Escors and Brecpot, 2010). The translational
mechanism of the host synthesizes and modifies viral proteins. Newly synthesized viral
proteins and full-length RNAs combine to produce a new virus form, which will be budding
from the host cell (Hu and Pathak, 2000).
A retrovirus infects the target cell by providing interaction between viral envelope protein
and cell surface receptor on the target cell. The virus then internalizes to the place where its
single-stranded RNA turns into double-stranded DNA. Double-stranded DNA is delivered
to the nucleus and integrated to the host cell genome there. Many types of retrovirus types
require degradation of mitosis and then the nuclear envelope for the arrival of a viral
genome within the nucleus (Robbins and Ghivizzani, 1998).
Stable binding of viral DNA to the host genome is advantageous because it will provide
long-term expression of transgenes required for therapeutic effect. However, one of the
disadvantages of current retroviral transfer systems is that they are not specific to types of
target cells (Yi et al., 2011).
One of the most important properties of retroviruses is that they can integrate a reverse
transcribed genome to the host cell chromosome (Miyazaki et al., 2011).
Unlike adenoviral vectors, retroviral vectors realize transfection through transgene
integration to the target cell genome. However, transgene expression ceases within a few
days or weeks. Retroviral vectors reveal gene transfer that is unsuitable for many cells in in
vivo conditions, and this partly stems from rapid inactivation of the human complement
system. This silencing is not well characterized; however, it appears as a result of
methylation in DNA or promoter regions, or participation of splice region to condensed
chromatin (Reynolds et al., 1999; Navarro et al., 2008).
The ability of retroviruses to enter their genomes to host DNA enables them to make stable
modifications to the life cycle of the host cell. This stability is greater than viruses such as
adenovirus, herpes simplex virus (HSV), and papilloma virus, which remain episomal
(Robbins and Ghivizzani, 1998).

2.2.1.3. Lentiviral systems


In recent years, studies mainly concentrated on the use lentiviral vectors. Lentivirals are
viral systems without small, retrovirus-like viral proteins and no capacity for replication;
448 Recent Advances in Novel Drug Carrier Systems

they provide gene delivery to non-dividing cells. This characteristic property is an


advantage for various gene therapy applications used in targeting in post-mitotic and highly
differentiated cells. The most important advantage of lentiviruses compared with other
retroviruses is their ability for gene transfer to non-dividing cells (Escors and Brecpot, 2010;
Yi et al., 2011; Matrai et al., 2009; Freed, 1997). This characteristic is believed to be related to
the pathogenic characteristics of lentiviruses, which infect terminally differentiated cells of
monocyte/macrophage lineage (Freed, 1997). For this reason, lentiviral vectors can be used
for transgene expression to neuron cells (Yi et al., 2011).
Genome of lentiviruses have a more complicated structure; they contain accessory genes
which regulate viral gene expression, control combination of infectious particles, modulate
viral replication in infected cells, and are associated with the continuance of infection
(Howarth et al., 2010). In other words, the fact that it contains regulatory and accessory
genes apart from gag, pol and env, requires lentiviruses to adopt a more complex form
(Osten et al., 2007). For example, HIV-1 is one of the most widely used lentiviral vectors, and
contains six accessory genes (tat, rev, vif, vpr, nef, vpu) (Osten et al., 2007; Yi et al., 2011).
These proteins are involved in all steps of cell cycles, which are termed: budding,
maturation, and integration. In addition to helping combination of virions, these proteins
ensure nuclear export and transcription ratio of RNA (Yi et al., 2011).
HIV infects CD4-positive T lymphocytes and macrophages (CD4 antigen acts as a primer
surface receptor for HIV-1), and causes chronic immune deficiency, known as acquired
immune disorder syndrome (AIDS). HIV-1 genome additionally contains 2 regulatory genes
(tat and rev), and 4 accessory genes (vpr, vpu, vif, nef). The lenti viral genome is 9.2 kb in length.
Tat protein increases transcriptional activity in 5′ LTR of integrated provirus from promoter
regions. Rev protein transports non-spliced HUV mRNA from the nucleus. In general,
accessory proteins increase virulence against the host organism. Development of HIV-1-based
vectors was first achieved by utilizing the abilities of lentiviral vectors of infecting post-mitotic
cells. The design of other retrovirus-based vectors, such as self-inactivating MoMLV vectors,
can be directly transferred to lentiviral vectors. In the most recent generations of HIV-1-based
vectors, all accessory genes and the regulatory tat gene are deleted (Osten et al., 2007). In the
self-inactivating (SIN) expression vector, the U3 promoter region is deleted from the 3’LTR.
This region was copied to the 5′ end of dsDNA during reverse-transcription. Therefore, SIN
modification results in transcriptional inactivation of the integrated provirus, which allows for
the restriction of the likelihood of recombination with latent retroviral sequences and
mobilization of latent retroviral sequences in the host cell genome (Osten et al., 2007).
Tropism of lentiviral vectors depends on the type of envelope protein used for viral
production (Escors and Brecpot, 2010). The use of heterologous Env proteins is called
pseudo typing. The most widely used Env protein is “vesicular stomatitis virus glycoprotein
(VSV-G)”. This protein permits the virus high titration values via ultracentrifugation and a
wide tropism (VSV-G binds to suitable phospholipid component of plasma membrane)
(Osten et al., 2007).
Lentiviral vectors do not require degradation of the nuclear membrane for integration.
Lentiviruses that are encoded with the Gag matrix protein integrase enzyme and vpr protein
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 449

interact with the nuclear import mechanism of the target cell and manage active transport of
pre-integration complex via nucleopores. This characteristic enables transfection to non-
dividing cells and, in conclusion, “complex retroviruses” become suitable for use as gene
transfer vectors, particularly for post-mitotic cells like neurons (Howarth et al., 2010).
Receptors have been defined for many retroviruses. The best-characterized example is CD4
molecule, which serves as a receptor for lentiviruses including HIV (Freed, 1997).

Frequently, pseudotype lentiviral vectors are formed by “vesicular stomatitis virus”


envelope (VSV-G). VSV-Gis a glycoprotein that interacts with the phospholipid component
of a number of receptors or cell membrane. VSV-G offers large host-cell range and high
vector particle stability, which are attractive characteristics for ex vivo gene modification.
However, restriction of infection to specific cells, which is called transduction altargeting, is
effective and reliable and critical in the case of in vivo gene delivery. In addition, it is the key
to increase therapeutic effectiveness, reduce adverse effects, and reducing the required
amount of the vector. Two methods can be used to achieve this: (a) benefiting from natural
characteristics of existing viral proteins, (b) using gene engineering to continue, stop or
increase the original tropism of the vector (Escors and Brecpot, 2010).

Recently-developed lentiviral gene transfer systems have many characteristics of retroviral


systems. A viral genome integrates with host chromosomes, and genes that are desired to
remain permanently are placed (Yi et al., 2011).

2.2.2. Non-viral gene delivery systems


The basic concept underlying gene therapy is that it develops gene expression to specific
cells in order to treat human diseases or for transfer of genetic material to inhibit the
production of a target protein (He et al., 2010).
In theory, viral carriers can achieve rapid transfection of foreign material spliced to viral
genome with high transfection ratio. However, many studies that used viral vectors
reported unsatisfactory results, due to the immunologic and oncogenic adverse effects of
these vectors. On the other hand, non-viral vectors have many advantages, such as easy of
fabrication, cell/tissue targeting, and low immune response. The biggest disadvantage of
non-viral vectors in clinical use is low transfection efficiency (He et al., 2010). Therefore, the
most important points that should be taken into account in gene therapy are the
introduction of the gene into the cell and increasing transfection efficiency. Using
transfection, cells are induced for the production of chemicals used in pharmacological
arrangements like hormone replacement or specific protein production (Godbey and Mikos,
2001). Naked DNA molecules cannot effectively enter to the cell due to their hydrophilic
structures and large size resulting from negatively-charged phosphate groups. In addition,
they are easily fragmented by nuclease enzymes. Therefore, the biggest difficulty in gene
therapy is the development of physical methods to ensure gene transfer to target cells of the
gene delivery vectors and delivered gene (Al-Dosari and Gao, 2009).

As seen in Figure 4, natural and synthetic polymers are used to prepare non-viral gene
delivery systems. Compared to viral delivery systems, non-viral carriers are less toxic and
450 Recent Advances in Novel Drug Carrier Systems

immunogenic. Another advantage of non-viral vectors is ease-of-production (DeLaporte et


al., 2006). Much of the previous research found that non-viral systems were less effective
than viral systems. However, in gene therapy conducted with physical methods, the
effectiveness of gene transfection and therapy increased, and the duration of gene
expression was extended significantly in clinical terms (Al-Dosari and Gao, 2009).

Figure 4. Modular design of non-viral vectors schematic. Modules associated with vector design are:
vector backbone (grey), functional groups for regulating environmental interactions (purple), and
intracellular trafficking (red). The vector backbone, typically containing polymers, lipids, or
polysaccharides, is designed for DNA binding and complexation, which can protect against nuclease
degradation, create a small, less negatively charged particle that can be internalized by cells, and facilitate
some intracellular trafficking. The function of the vector backbone is being augmented by the attachment
of groups that address the extracellular and intracellular barriers. The environmental functional groups can
serve to limit interactions with serum components, promote specific cell binding or tissue targeting, or
facilitate interactions with the extracellular matrix or biomaterials. The intracellular functional groups aim
to enhance nuclear accumulation of the DNA either by facilitating endosomal escape, movement along the
cytoskeleton, or nuclear pore trafficking. The individual modules can be assembled in different ways (a–c)
for complexation with DNA (green), which may affect the structure and function of the resulting non-viral
vector. (d) Schematic illustrating the distribution of the modules and DNA throughout the vector cross
section, with the desired organization of functional groups regulating the environmental interactions
presented primarily on the exterior and the groups for intracellular trafficking protected within the vector
interior for activity following internalization. (e) Vectors are internalized by endocytosis and must
subsequently escape the endosome for transport to the nucleus. Additionally, the modular components
must dissociate from the DNA to allow for transcription (DeLaporte et al., 2006).
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 451

A number of barriers need to be overcome in order to increase the effectiveness of non-viral


vectors in humans. These barriers are classified as production/formulation/storage;
extracellular barriers; and intracellular barriers (Davis, 2002). Anatomic barriers are
extracellular matrixes coating the cells, which prevent direct transport of macromolecules to
target cells through epithelium and endothelial cell sequences. Phagocytes like Kupffer cells
in the liver and residential macrophages in the spleen are responsible for theclearance of
DNA-loaded colloidal particles in blood circulation. Similarly, nucleases found in blood and
extracellular matrix cause free and unprotected nucleic acids to be rapidly inactivated
following systemic application. The most critical barrier to effective DNA transfection was
regarded as the transition of plasma membrane. Typically, naked nucleic acids cannot cross
cell membrane by cellular uptake mechanisms such as endocytosis, pinocytosis, and
phagocytosis without application of physical methods or being loaded to a carrier (Al-Dosari
and Gao, 2009). There is a substantial body of research on developing effective physical,
chemical, and biological systems to deliver transgenes into the cell to provide appropriate
expression (Gao et al., 2007). Physical approaches, including electroporation, gene gun,
ultrasound, and hydrodynamic delivery are based on the application of a force to increase the
permeability of the cell membrane and promote intracellular gene transfer. In chemical
applications, synthetic or natural carriers are used to transport transgenes to the cells. An ideal
gene delivery system should meet 3 criteria: The carriers should protect the transgene from
nuclease enzymes inside intracellular matrixes; should transport the transgene from plasma
membrane to the target cell nucleus; and should not cause any toxic effect (Gao et al., 2007).
Two of the most important advantages of synthetic carriers are that they do not display
immunogenicity, and large-scale production is easy. Furthermore, the size of the gene to be
delivered does not reduce efficiency of these systems. Even mammalian artificial
chromosomes of 60 megabase were successfully transfected by these types of carriers
(Schatzlein, 2003).

Non-viral vectors can trigger an inflammatory response, since they do not provide a specific
recognition; they are much less dangerous than viral vectors in terms of antigen specific
immune response. However, although non-viral vectors seem to be more suitable, there are
some important points to be contemplated in vector design. Non-viral vectors should be
designed according to specific cell targeting; cellular uptake and cellular release should be
optimized; and potential immune response should be minimized (Conwell and Huang,
2005; Mrsny, 2005).

2.2.2.1. Physical methods

Gene Gun
Delivery with gene gun method is also termed ballistic DNA delivery or DNA-coated
particle bombardment, and was first used for gene transfer to plants in 1987 (Al-Dosari and
Gao, 2009; Lin et al., 2000). This method is based on the principle of delivery of DNA-coated
heavy metal particles by crossing them from target tissue at a certain speed. The particles
achieve sufficient speed due to a pressurized inert gas (generally helium). Generally, gold,
tungsten or silver microparticles were used as the gene carrier (Mhashilkar et al., 2001; Al-
452 Recent Advances in Novel Drug Carrier Systems

Dosari and Gao, 2009; Miyazaki et al., 2006). Momentum allows penetration of these
particles to a few millimeters of the tissue and then cellular DNA release (Gao et al., 2007).
These particles typically have a diameter of 1µm. Due to its small size, the particle easily
penetrates the cell membrane and can transport DNA into the cell. At this point, DNA
separates from the carrier particle and can be expressed (Lin et al., 2000).

Gas pressure, particle size and dose frequency are critical factors in determining the degree
of tissue damage and penetration effectiveness of the application (Al-Dosari and Gao, 2009).

Gene-gun-based gene transfer is a widely tested method for intramuscular, intradermal and
intratumoral genetic immunization (Miyazaki et al., 2006). Numerous animal tests and
clinical trials indicated that this method caused a greater immune response than
microinjection, even in low doses (Al-Dosari and Gao).

The advantages of gene gun over other in vivo gene delivery systems are as follows: [1] It
does not use toxic chemicals or complex biological systems, [2] delivery is achieved without
the need for a receptor, [3] DNA fragments of various sizes, including large ones, are
transported, [4] there is no need to introduce foreign DNA or protein, [5] it has high
repeatability, [6] production of heavy metal particles is easy (Lin et al., 2000). However, in
this method, gene expression is short-term and low (Mhashilkar et al., 2001).

Electroporation

The future of gene therapy requires the advancement of effective and non-toxic
polynucleotide gene delivery mechanisms. Electroporation includes controlled electric
application to increase cell permeability. Electroporation was first developed in 1960s, with
studies on the degradation of cell membrane with electric induction. Neumann et al., first
reported transfection of eukaryotic culture cells through electroporation in 1982. In many
subsequent studies, transfection was performed on animal and plant cells via
electroporation (Al-Dosari and Gao, 2009; Somiari et al., 2000).

Electroporation introduces foreign genes into the cell by electric pulses. In this method,
pores are formed on the membrane surface to enable the DNA to enter the cell. Pore
formation occurs very rapidly, in approximately 10 nanoseconds. The size of the electric
pore is estimated to be smaller than 10 nm radius. If the molecule is smaller than the pore
size (as in oligonucleotides and chemical compounds), it can be transferred to the cell
cytosol through diffusion (Somiari et al., 2000; Miyazaki et al., 2006). In addition to passive
diffusion, loaded molecules and ions can be transported from the membrane via
electrophoretic and electro-osmotic means via the effect of electric regions. The pores close
following cellular uptake of DNA. In recent years, in vivo electroporation became common
in nucleic acid vaccines and improving non-viral gene therapy. There are critical steps
defined for transportation via in vivo gene delivery with electroporation. The first step is
fusing together, attachment or proximity between the cell membrane and nucleic acid. The
second step is adding nucleic acid to the membrane, which is immediately followed by the
third step, involving the delivery of nucleic acid from the membrane. The delivery
procedure results in decreased pore size and permeability (Somiari et al., 200).
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 453

In vivo electroporation technique is a generally reliable method that is more efficient than
other non-viral systems. When the parameters are optimized, this technique is equally
effective as viral vectors. In addition to local injection and electroporation, it was previously
shown that in vivo electroporation can be applied in localized form after the injection of
plasmid DNA (Al-Dosari and Gao, 2009).

There are some difficulties in in vivo applications of electroporation. There is an effectiveness


region of approximately 1 cm between the electrodes, and this makes transfection of the
cells in large regions of the tissues difficult. It is difficult to use in internal organs, and
surgery is required to implant electrodes. High temperature due to high voltage application
can cause irreversible tissue damage. The application of high voltage to cells can affect the
stability of genomic DNA (Al-Dosari and Gao, 2009; Gao et al., 2007).

Ultrasound

Ultrasound has many clinical advantages as a gene delivery system, due its easy and
reliable procedure (Pitt et al., 2004; Ferrara, 2010). In recent years, it was found that
microbubbles applied by ultrasound increased gene expression. Microbubbles or ultrasound
contrast agents decrease cavitation threshold with ultrasound energy. In many applications,
perfluoropropane-loaded albumin microbubbles (Mallinckrodt, San Diego, USA) were used.
The microbubbles were modified with plasmid DNA before the injection and then
ultrasound was applied (Niidome and Huang, 2002).

Ultrasound has been used for therapeutic purposes for more than a half century, even before
the use of ultrasound for diagnosis and imaging purposes. In diagnostic use, low-intensity
ultrasound is used to prevent energy accumulation in tissue causing biological effects.
Conversely, therapeutic applications are based on the principle of intensification of
ultrasound energy within the tissue (Frenkel, 2008).
After the discovery of the applicability of ultrasound to gene therapy at the cellular and
tissue level, this was taken one step further with physical methods. Reporter gene
expression, where naked DNA is used, advanced for10 times thanks to this discovery. The
transfection efficiency of this system is based on frequency, time of ultrasound treatment,
the plasmid DNA mount used, etc. In conclusion, size and local concentration of DNA plays
an important role in the effectiveness of transfection (Gao et al., 2007).

Hydrodynamic therapy

Hydrodynamic gene delivery is based on the principle of understanding the characteristics


and structure of capillaries, an understanding of the dynamic characteristics of the fluids
passing through blood veins (Suda and Liu, 2007). Hydrodynamic gene delivery uses the
hydrodynamic pressure created by the injection of the large volume of DNA solution with
blood pressure inside veins (Suda and Liu, 2007), and thus the permeability of the capillary
endothelium increases and pores form in the plasma membrane encircling parenchyma cells
(Gao et al., 2007; Suda and Liu, 2007). DNA or other related molecules can reach the cell
from these pores. Membrane pores are later closed, and these molecules are retained within
the cell. In hydrodynamic gene therapy, the principle reason for targeting parenchyma cells
454 Recent Advances in Novel Drug Carrier Systems

is that capillary endothelium and parenchyma cells are closely related, and when the
endothelial barrier is breached, this allows DNA to easily reach parenchyma cells. In
addition, the capillary’s thin wall structure has a stretchable and easily fragmented structure
(Suda and Liu, 2007). The effectiveness of hydrodynamic procedure depends on capillary
structure, the structure of the cells encircling capillaries, and the hydrodynamic force
applied (Gao et al., 2007; Suda and Liu, 2007).

2.2.2.2. Chemical methods

Polymers
Polymers are long-chained structures composed of small spliced molecules called
monomers. Polymers that are composed of a repeated monomer are called homopolymers,
while those composed of two monomers are called copolymers. Natural and synthetic
polymers are used in drug delivery systems. Biodegradable and non-biodegradable
polymers are used according to the type of controlled release mechanism. Biodegradable
polymers are non-water soluble, and undergo chemical or physical change in biologic
environments (Tuncay and Calis, 1999). Polyamides, dextran, and chitosan are examples of
biodegradable polymers. On the other hand, non-biodegradable polymers are not degraded
in biological environments; hydrophilic polymers are hydrogels, which are non-water
soluble and swell in water, while hydrophobic polymers are non-water soluble and do not
swell. Examples of hydrophilic hydrogel polymers include polyvinylalcohol,
polyvinylacetate, polyethyleneglycol, polyacrylic acid, polyhydroxyethyl methacrylate, and
polymethacrylic acid. Examples of hydrophobic polymers include silicones, and
polyethylene vinyl acetate. Polymers such as ethyl cellulose (EC), hydroxypropyl methyl
cellulose (HPMC), cellulose acetate phthalate (CAP), and eudragit derivatives are commonly
used in controlled release systems (Tuncay and Calis, 1999).

Selection and design of a polymer is difficult due to structural differences; achieving the
desired chemical, mechanical, and biological functions requires understanding of the
characteristics and surface of a polymer. For polymer selection, in addition to its
physicochemical characteristics, characterization of extensive biochemical characteristics and
preclinical tests are required to demonstrate its reliability (Pillai and Panchagnula, 2001).
The physical properties of a polymer carrier, such as hydrophilicity, surface charge,
permeability and diffusibility, biocompatibility with tissue and blood (Pillai and
Panchagnula, 2001).
For successful delivery, polymers should package DNA in small sizes. Thus, extracellular
and intracellular stability of DNA is increased; cellular uptake by endocytosisis enabled;
and, by transporting it to the nucleus, the active form of DNA can be released within the
nucleus (Mrsny, 2005).

Polymeric gene transporters deliver genetic material through electrostatic interactions with
nano-sized polyplexes for gene therapy (Kim et al., 2011).

A synthetic gene delivery system: [1] should protect the negatively-charged phosphate DNA
skeleton against anionic cell surface from load repulsion, [2] should be condensed to suitable
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 455

length intervals of DNA with a macromolecular structure for cellular internalization


(condensing at nanometer size for receptor compatible endocytosis or condensing at
micrometer size for phagocytosis); and [3] should protect DNA from all extracellular and
intracellular nuclease degradations. To meet this need, researchers concentrated on three
strategies: electrostatic interaction, encapsulation, and adsorption (Wong et al., 2007).

Electrostatic interaction

The majority of polymeric vectors presently in use form complexes with negatively charged
DNA by electrostatic interaction. All polymers have amino groups. At adequate nitrogen–
phosphate ratio, the polymer and the DNA form nanocomplexes, which allows both cellular
DNA uptake and also protects the DNA from nuclease enzyme (Wong et al., 2007).

Encapsulation

An alternative to electrostatic condensation of DNA is encapsulation of DNA with a


biodegradable polymer. Polymers that have an ester linkage in their structures (like
polyesters) are hydrolytically degraded to short oligomeric and monomeric compounds,
which are more easily discharged from the body. Furthermore, the degradation mechanism
and DNA release can be controlled by changing the physicochemical characteristics and
composition of the polymer. DNA is protected from enzymatic degradation by
encapsulation (Wong et al., 2007) (Figure 5).

Figure 5. Gene packaging—The three main strategies employed to package DNA are via (1) electrostatic
interaction, (2) encapsulation within or (3) adsorption onto biodegradable nano- or microspheres (Wong
et al., 2007).
456 Recent Advances in Novel Drug Carrier Systems

Adsorption

Alternative DNA packaging methods, such as adsorption techniques, were developed to


overcome the limitations of encapsulation methods. DNA adsorption represents the
combination of the above mention two models. In this method, DNA is conjugated to the
surface of biodegradable cationic particles, or is electrostatically adsorbed due its negative
charge. This method prevents the DNA from being exposed to heavy encapsulation
conditions, and also increases rapid release DNA (Wong et al., 2007).

Adsorption methods attracted much research interest, as it does not cause immunogenicity,
does not require the integration of exogenous genes to host chromosomes, is a simple and
cheap method, and is suitable for effective gene delivery. However, low transfection
efficiency, some cytotoxic effects, and in vivo instabilities are still significant restricting
factors in gene therapy. One of the strategies to solve these problems is the synthesis of
biologically degradable polymers (Kim et al., 2011).

Liposomes

Liposomes are colloidal drug delivery systems. They are biologic membrane-like sacs in
sphere form, formed by one or more lipid layers, and include an aqueous phase. The
phospholipid phase consists of principle components like aqueous phase and cholesterol.
Liposomes are classified according to the number of layers they contain. The advantages of
liposomes include effectiveness at small doses, extended dosing interval, and ideal transport
for active substances with a short half-life. Cellular uptake mechanism of active substances
in liposomes can be categorized as endocytosis, combination by melting, and adsorption.
Liposomes are used for application of carcinogenic, antifungal, antiparasitic, antiviral, and
anti-inflammatory drugs, hormones, DNAs, and cosmetics (Bogdansky, 1990).

Cationic lipids, which are used for the entrance of plasmid DNAs in the cell, have been
studied for more than 20 years. During this period, many cationic liposome formulations
were developed, and were tested as nucleic acid transported in animals and human with
phase I and phase II studies. When compared to other gene delivery systems such as viral
vectors and transfection agents, cationic liposome delivery systems are more easily
formulated and cause no biological damage like viral vectors (Dass and Choong, 2006).
Critical parameters that determine the behavior of liposomes in in vitro and in vivo
conditions include size, number of layers, and surface charge. Unilamellar or mutilamellar
vesicles can be produced depending on the preparation method; the diameter of these
vesicle ranges from 25 nm to 50 µm (Bogdansky, 1990).

Liposomes are divided into three categories according to their charges: cationic, anionic, or
neutral. Compared to viral vectors, liposome delivery systems are non-pathogenic and non-
immunogenic, with easy of preparation. However, the most important disadvantage of
liposomes is short gene expression time and low transgene expression level. Since liposomes
are generally non-toxic and non-immunogenic structures, they are considered reliable
carriers for gene therapy. Liposome-based gene delivery was first reported by Felgner
(1987). In the 1990s, liposomes were used in numerous cationic lipid gene therapy tests.
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 457

Liposomes are not as effective as viral vectors in in vivo gene delivery, and it is generally
difficult for them to transfect targeted gene to selected cells (Miyazaki et al., 2006).

Dendrimers

Dendrimers were first defined in the late 1970s and early 1980s. Dendrimers consist of
symmetrical branches projecting from a central core (Dufes et al., 2005; Genc, 2008; Bulut
and Akar, 2012). Dendrimers are 10–200 Angstrom in diameter; they are repeating,
branched, large spherical molecules, and have functional groups on their surface that can be
used as a building block to connect many biological materials (Ward and Baker, 2008). These
functional groups determine the variability of dendrimers, whereas branching provides the
growth of the structure (Dufes et al., 2005; Bulut and Akar, 2012). As seen in Figure 6, the
most important advantages of dendrimers are conjugation of a large number of different
molecules on the dendrimer surface, and the conjugation of molecules that will be used for
diagnosis and treatment (Ward and Baker, 2008; Dufes et al., 2005).

Figure 6. Dendrimer structure. The stepwise synthesis of dendrimers means that they have a well
defined hierarchical structure. This hypothetical dendrimer is based on a core with three covalent root
attachment points but other common cores have di- or tetracovalent cores. The valency of the core
dictates the number of linked dendrons and the overall symmetry of the molecule. The dendrons are
synthesised by covalent coupling of the branch units. For each additional layer or generation that is
being added to the structure the reaction sequence is repeated. In this case the units have two new
branching points at which additional units can be attached. (The generation count is not always
consistent: normally generation 0 refers to the core while sometimes it is used to describe the dendrimer
after the first reaction cycle.) The number of branching points, branching angles, and the length of the
branching units determine to what extent each generation increases molecular volume vs. surface area.
For the higher generations the density of the terminal groups reaches a point where for steric reasons no
groups can be added (starburst effect). Dendrimers of higher generation also have a typical molecular
density profile under favourable conditions; the high peripheral molecular density establishes a steric
outer shell and the lower density at the centre creates cavities which can accommodate guest molecules.
(Dufes et al., 2005).

The degree of polymerization of dendrimers is represented by the number of generations


(G) of repeated branching cycles during synthesis. Generation number can be easily
identified by calculating number of branching points from the core to the outer surface.
458 Recent Advances in Novel Drug Carrier Systems

The number of branching points increases proportional to dendrimer growth. For


example, a dendrimer with no connection points is termed zero generation (G-0) (Bulut
and Akar, 2012).

Equal layer groups of dendrimers with an equal number of layers are highly suitable for use
in drug delivery due to their perfect encapsulation characteristics and highly controllable
chemical behaviors. Drugs can be encapsulated in a dendrimer or can electrostatically or
covalently connect to functional groups on the surface (Ward and Baker, 2008; Bulut and
Akar, 2012). On the other hand, targeted drug delivery can be achieved by covalent
conjugation of the targeting agent to functional groups of dendrimers (Ward and Baker,
2008).

Although the toxicity of dendrimers that form complexes with DNA generally decreases,
toxicity and cellular effects of free dendrimers should be taken into account (Dufes et al.,
2005).

Cationic lipid compatible systems

Felgner et al. pioneered cationic lipid-based gene transfer (Felgner et al., 1987; Al-Dosari and
Gao, 2009). Today, hundreds of lipids have been developed and tested for gene transfer.
Lipid-based systems consist of a positively-charged hydrophilic head and hydrophobic tail
structure.

Most common hydrophilic head groups are primary, secondary, tertiary amines, or
quaternary amine salts. These positively charged sections connect to negatively charged
phosphate groups in nucleic acid. The hydrophobic tail consists of aliphatic chain,
cholesterol, or steroid rings. The connection region between hydrophilic and hydrophobic
sections consists of the bonds that affect biodegradability ratio, such as ether, ester,
carbamate, or amid (Al-Dosari and Gao, 2009; Uyechi-O’Brian and Szoka, 2003).

Each lipid has a different sized head group and hydrocarbon tail length, and these
characteristics have a significant impact on the formation of DNA–lipid complexes and
intake by the cell (Balasz and Godbey, 2010). The transfection effectiveness of cationic lipids
varies according to the structure of the lipid and the charge ratio used to form the DNA–
lipid complex. Compared to negatively-charged DNA, a positively-charged lipid
spontaneously forms structures called lipoplexes. In lipoplex structure, the DNA molecule is
encircled by positively-charged lipids, which protect it from extracellular and intracellular
nucleases. In addition, due to their positive charges, lipoplexes electrostatically interact with
the cell membrane and thus cellular uptake is achieved (Al-Dosari and Gao, 2009; Uyechi-
O’Brian and Szoka, 2003).

Cationic lipids such as N-[1-(2,3-dioliloksi)propyl]-N,N,N-trimethlammoniumchloride


(DOTMA), [1,2-bis(oliloxy)-3-(trimethlamonnium) propane] (DOTAP), 3β[N-(N_,N_-
dimethlaminoethane)-carbamile] cholesterol (DCChol) and dioctadecylamidolysisspermine
are used to prepare lipid carriers (Balasz and Godbey, 2010).
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 459

The aim of cationic lipid-based gene therapy is to transport plasmid DNA to the cell and
thus to achieve the desired protein/peptide transcription and translation. DNA–lipid
complex is internalized into the cell through the vesicular path, which is followed by the
release of cell cytoplasm of DNA from the lipoplex. Some fractions of the released DNA are
transported to the nucleus (Uyechi-O’Brian and Szoka, 2003). Strong DNA–cationic lipid
complexes alone are insufficient for gene transfer; however, DNA release before or after
transport to the nucleus is critically important (Khalid et al., 2008). Lipid–cationic lipid ratio,
cationic lipid–DNA ratio and DNA mount presented to the cell are important parameters
that significantly influence transfection efficiency (Hofland et al., 1996).

Although gene expression is provided by cationic lipids, for clinical benefit, the
effectiveness, expression time and degree of gene transfer were not found to be adequate.
The most significant disadvantages of cationic lipids are structural instability and
heterogeneity, inactivation in blood, low effectiveness, and weak targetability when
compared to other systems (Navarro et al., 2008).

Cationic lipid-based transfection systems are easy to prepare and reliable; they also have
low immunogenicity, and there is no need to classify the size of DNA which will be
transported (Mortimer et al., 1999). Many cationic lipid-DNA based systems are effectively
taken up by endocytosis; however, the mechanism of DNA release from endosome and
translocation to the nucleus are not well known (Mortimer et al., 1999).

2.3. The use of gene delivery systems in treatment


2.3.1. Cancer
Any change in the structure of genes that cause diseases is called mutation. In a mutated
cell, variations occur in the sequence of nucleotides. As a result, chemical substances that are
produced under the control of the gene have abnormal structure and function. If the gene is
associated with the growth, differentiation, or division of cells, then the mutation might
cause cancer (Wang et al., 2012). There are two main groups of genes responsible for formation
of cancer. These are oncogenes and tumor suppressor genes. Oncogenes are mutated versions
of normal genes that are responsible for controlling cell growth. After mutation, they compel
the cell to become a cancer cell. There are more than 100 known oncogenes, most of which
were identified in human tumors. The “ras gene” is the most widely known and most
commonly studied of these genes. After mutation, the ras gene initiates division and cancer
formation in cells with the “ras protein” it forms (Luo et al., 2012).

Tumor suppressor genes normally inhibit cell division and tumor development in cells. The
mutated forms of these genes cannot inhibit cell division, and instead cause tumor
formation. Among these genes, the RB and P53 genes are the most widely known. These
genes show their effect with p-RB protein and P53 protein, which are produced under their
control. Normally, these proteins inhibit DNA replication and cell division, and regulate
apoptosis. If the created genes are mutated, these proteins lose their activity and
460 Recent Advances in Novel Drug Carrier Systems

uncontrolled cell division occurs, which results in cancer development. It was indicated that
RB protein was inactive in approximately 40% of human cancers. Similarly, it was
found that P53 gene had an abnormal structure in 50% of all tumors (Morandell & Yaffe,
2012).

Gene therapy is based on the principle of replacing damaged genes in diseased cells with
corresponding healthy genes that were prepared outside the body. This approach is the
most difficult type of gene therapy. Successful treatment involves healthy genes, which are
prepared out side the body, targeting specific cells in the body via certain transporters
(carriers). Current technology has simplified the process of preparing healthy genes.
However, the subsequent process of targeting cells remains imperfect (Lagisetty & Morgan,
2012).

In addition to the correction of defective genes, gene therapy includes other treatment
principles. In a different method, cancer patients were injected “cytosinedeaminase”
enzyme, which is not normally produced in humans, via viral carriers,thereby allowing this
enzyme to be synthesized in tumor cells (Logue & Morrison, 2012). The drug 5-FC is injected
at high doses with no adverse effects; this is converted to 5-FU in tumor cells by cytosine
deaminase enzyme. Using this method, effective levels of drug activation are achieved
without damaging healthy cells.

In a similar method, carrier viruses injected with “thymidinekinase enzyme gene” found
in the herpes simplex viruses reach cancer cells and produce thymidine kinase enzyme in
these cells. This enzyme enables the drug ganciclovir, which has no adverse effects on
normal body cells, to be active in tumor cells and destroy cancer cells (Etienne-Grimaldi et
al., 2012)

Another treatment approach involves inhibition of oncogene proteins that were mentioned
above. Oncogenes cause cancer formation via the proteins they control. If the production of
these proteins is inhibited, their cancer-causing effects will be reduced. One example is the
farnesyltransferase enzyme, which plays a role in the formation stages of RB protein. It was
reported that new tumor formation was inhibited in patients who were injected with drugs
that inhibited the function of this enzyme (Logue & Morrison, 2012). Cancer treatment is the
most studied application of gene therapy. A large number of genetic changes occur by the
transformation of a normal cell to a neoplastic cell (El-Aneed, 2004).

Tumor suppressor genes eliminate cancer cells through apoptosis, whereas oncogenes
accelerate proliferation. For this reason, apoptotic genes and anti-oncogenes are frequently
used in cancer treatment. In addition to oncogenes and tumor suppressor genes,
chemotherapy and gene therapy were combined via a ‘suicide gene’ strategy. A suicide gene
encodes an enzyme that does not belong to mammals, and this enzyme converts non-toxic
product into active cytotoxic metabolite in cancer cells. Tumor suppressor genes, anti-
oncogenes, and suicide genes target cancer cells at the molecular level. Generally the genes
that encode cytokine revive immune response to cancer cells (Wang et al., 2012).
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 461

Transformation of a normal cell to a neoplastic cell occurs by multi-mutationary changes of


the cells at the genetic level. Due to the complicated nature of cancer, cancer gene therapy
involves many therapeutic strategies. These strategies can be analyzed according to two
categories: immunogenic and molecular (El-Aneed, 2004).

2.3.2. Immunologic approach


The human immune system has two pathways to respond to foreign antigens. The first
pathway includes antibodies secreted by B cells following activation of membrane
immunoglobulin via (B cell receptor)-antigen connection. Antibodies are water-soluble
proteins that move within the circulatory system to reach the target antigens. T cells, the
second immune pathway, directly interact with antibodies without secreting antibody. T
cells can produce multiple immune reactions, including cytotoxic effects (Lagisetty &
Morgan et al., 2012).

Cancer cells are immunogenic in nature with their cancer antigens, which are intracellular
molecules. Therefore, cellular immunity (T-cell mediated), is more important than humoral
immunity (B-cell mediated) (Restifo et al., 2012). Normal immune response is not sufficient
to eliminate tumor cells. The ability of cancer cells evade immune-system responses is
associated with secretion of immunosuppressive factors, down-regulation of antigen
expression or MHC (major histocompatibility complex) molecules, and co-stimulation
deficiency. One of the common genetic immunotherapy strategies includes transfer of
immune-stimulating molecular genes like cytokines. Immune response is created by the
activation of interleukin-12 production by tumor cells with numerous components of
immune system, particularly including cytotoxic T lymphocytes and natural killer cells
(McDonald et al., 2012).

Another immunologic approach involves in vitro manipulation of the antigen-presenting


cells to produce active tumor antigens. Dendritic cells are the strongest antigen-presenting
cells (El-Aneed, 2004). Application of antigen-encoding genes via direct vaccination might
induce the desired immune response to cancer cells. When injected subcutaneously or
intramuscularly, DNA enters local cells like fibroblast and myocytes, and antigen is
subsequently produced and secreted. Antigen-presenting cells move towards lymphoid
organs where new antigens will be caught and the desired immune response will be
initiated (El-Aneed, 2004). Another method to increase the specificity of chemotherapy
might be enzyme-encoding enzyme-activate prodrug therapy, which converts non-toxic
products into specific and toxic metabolites (Altaner, 2008). This approach is known by
many different names, including enzyme prodrug therapy in gene management; suicide
gene therapy or enzyme prodrug therapy in virus management; and gene prodrug
activation therapy (Scheier et al., 2012). All of these expressions define the same two-step
treatment process. The first step involves the targeting and transport of the foreign
enzyme to the tumor via various pathways. In the second step, non-toxic prodrug is
applied and converted to active systolic metabolite. In in vivo approaches, expression of
foreign genes does not occur in each cell of the target tumor. As a result, the active drug
462 Recent Advances in Novel Drug Carrier Systems

can enter the transduced cell and cell deaths take place, including the cells which do not
express enzyme. This procedure is also termed the bystander effect or neighbor cell death
effect. In addition, dead cells can induce host immune response due to T cells and NK
cells. This therapeutically beneficial effect is known as the slight bystander effect. To
increase the effectiveness of enzyme-activated product therapy, therapeutic genes in the
vector should be expressed at a sufficiently high ratio to achieve effective prodrug
transformation; it should remain as active as possible in order to support the killing
mechanisms of the produced systolic metabolite, and the suicide gene should be
specifically targeted only to the tumor. These arrangements will also reduce adverse
effects (Altaner, 2008).

2.3.3. Cardiovascular disorders


Recombinant adenovirus is one of the most practical vectors for localized gene therapy. In
addition, these vectors allow for tissue-specific delivery, and are suitable for re-targeting.
Bispecific antibodies were used to coat adenovirus particles by connecting the antibody
ligand on the cell receptor surface. Genetic modifications also include gene modifications for
some viral coat proteins to change tropisms of adenoviruses (Ji et al., 2012).

Antiviral vectors are the most recently developed vectors; they are HIV-based and generally
used for HIV treatment. These vectors have the ability to transfect non-dividing and
dividing cells. The effectiveness and reliability of these cells are currently being analyzed
(Kozarsky, 2001).

2.3.3.1. Angiogenesis

One exciting recent development is that blood circulation can be provided to ischemic
tissues. The development of new blood veins was observed in the region of endothelial
growth factor (VEGF) expression (Katz et al., 2012). Gene transfer has advantages for protein
therapy, and the use of permanent VEGF is preferred. This method can be fulfilled by a
single injection of gene-transfer vector; however, transgene expression of VEGF should last
for a limited period, because long-term unregulated VEGF expression might cause abnormal
angiogenesis. Therefore, rapid gene expression obtained from adenoviral and plasmid
vectors are among the ideal vectors for local delivery of VEGF gene by direct injection
(Kozarsky, 2001; Katz et al., 2012).

2.3.3.2. Cardiac failure

Treatment of cardiac failure by gene therapy is still limited, due to the need to determine
suitable transgenes and develop effective gene delivery methods to the myocardium. Genes
can be directly transported to the heart. However, if these genes have to be transported to
larger regions, as in the case of cardiac failure, this method is not regarded as successful.
Another method involves transporting gene delivery vectors to coronary veins via a
catheter; however, some catheter materials inactivate recombinant adenovirus (Kozarsky,
2001; Kairouz et al., 2012).
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 463

2.4. Pulmonary disorders


According to some authors, lungs are ideal organs for gene therapy. However, cystic fibrosis
and α1-antitripsin deficiency are the only common disorders with known genetic
background for which existing treatments are not curative. Although cystic fibrosis is the
first target for pulmonary gene therapy, this method is used to treat many acquired diseases,
mainly cancer (Davies et al., 2003).

After the discovery of CTFR in 1989, considerable new progress was made to improve gene
transfer system for somatic gene therapy of cystic fibrosis. Cystic fibrosis affects
approximately1 in 2500 newborns, which makes it the most common recessive genetic lethal
disorder (Davies et al., 2003). Even a copy of a single normal SF gene is sufficient for normal
function (Klink et al., 2004). Therefore, somatic gene therapy is relatively straightforward.
The only requirement is to provide a cell affected by a gene which makes CFTR protein
expression (94). CFTR is cAMP-regulated chloride channel in epithelium cells. Dysfunction
of CFTR arises from production deficiency, problems in transport of apical membrane of the
cell to the effective region, or due to functional defects (Davies et al., 2003). Air tract
epithelium is the most important target among lung diseases that cause mortality and
morbidity. In order to address this aim, many gene delivery systems were developed and
their effectiveness in vitro and in vivo was analyzed.

The use of adenoviral vectors gave rise to safety concerns due to low transduction
effectiveness and increasing inflammation in human lungs. Tests with cationic liposomes
showed low toxicity and effectiveness (Klink et al., 2004).

Some individuals have to be overcome to perform gene therapy. Mucociliary clearance is the
primary one of these individuals. Air tract secretions and the mucociliary clearance system
act as significant barriers to exogenous gene transfer. The epithelia of a healthy respiratory
system create a thin mucus layer required for normal cilia function; however, this layer
inhibits gene transfer by cationic liposomes (Davies et al., 2003).

Similarly, a potential gene transfer agent has to overcome the barrier formed by secretions in
order to enter the cell. The glycocalyx structure and endocytosis ratio of a cell were defined
as the factors that restrict the effectiveness of gene transfer. A number of intracellular
process, including endosomal system, cytoplasmic delivery, and the entrance to the nucleus
serve as barriers to successful gene transfer. Similarly, host immune response causes
problems in the application of gene transfer agents to lungs (Davies et al., 2003).

Adenovirus/CFTR was applied to the nasal epithelia of three patients to treat cystic fibrosis.
No viral replication was observed, but mild inflammation was reported. PD returned to
normal baseline values in all three patients (Davies et al., 2003). However, one of the
problems of this vector system is that low expression levels of viral gene wastes leads to
cytotoxic T-cell response that targets vector-containing cells. The fact that adenoviruses are
generally air tract pathogens is a both advantageous and disadvantageous. Many potential
464 Recent Advances in Novel Drug Carrier Systems

receivers have circulating antibodies that reduce vector effectiveness and T-cell response.
Most importantly, systemic application of adenoviral vectors causes acute and potentially
life-threating inflammation responses due to activation of antigen-presenting cells (Klink et
al., 2004). On the other hand, when compared to other gene delivery systems, lentiviral
vectors are advantageous. Lentiviral vectors are integratable retrovirus derivatives with a
high cloning capacity. When suitable regular sequences such as LCS (locus control sections)
are used, stable and cell-specific expression can be achieved. When envelope proteins of
pseudo-written lentiviral vectors are applied to apical surface with vesicular stomatitis
virus, it is not possible to effectively transduce polarized air tract epithelia. Retroviral
vectors, pseudo-written with apical membrane-connecting envelope proteins, are used to
overcome this problem (Klink et al., 2004).

The first clinical trial of liposome-compatible CFTR gene transfer was reported in 1995. This
method involves direct application of DC-Chol/DOPE/CFTR to the nasal epithelia of SF
patients. No adverse clinical effects were observed, and nasal biopsy analyses showed no
histologic change. Specific mRNA and plasmid DNA were found in five out of eight
patients who received CFTR gene vector (Klink et al., 2004).

3. Conclusion
Current gene delivery systems are divided into three categories: viral-based, non-viral
based, and combined hybrid systems. Viral gene delivery systems consist of viruses that are
modified to be replication-deficient which can deliver genes to the target cells to provide
expression. Although viral systems have many advantages, such as long-term expression,
and the effectiveness and expression of therapeutic genes, they also have various
disadvantages. These disadvantages include risks due to working with viruses, optimization
problems when producing large batches, and problems of immunogenicity and toxicity. To
date, adenoviruses, retroviruses, and lentiviruses have been used as viral gene carrier
vectors. Non-viral systems include physical and chemical methods. In its broadest sense,
this refers to delivery of the gene into the cell by applying a physical force in order to
increase the permeability of the cell membrane. The most common physical methods are
microinjection, electroporation, ultrasound, gene gun, and hydrodynamic delivery.
Chemical methods use natural and synthetic compounds are to deliver the gene into the cell.
The gene delivery system generally includes polymers, liposomes, dendrimers, and cationic
lipids. Non-viral vectors have many advantages, such as easy of synthesis, effective
targeting of cell/tissue, low immune response, and potential to use plasmid at desired
molecular weight. The biggest disadvantage of non-viral vectors in clinical use is low
transfection efficiency. A large number of viral and non-viral gene delivery systems have
been developed. Both systems have many advantages and disadvantages. Hybrid systems
were developed to combine the advantageous parts of both the individual systems, while
minimizing their respective disadvantages, thereby producing a gene therapy with higher
effectiveness and low toxicity. The biggest advantage of hybrid systems is that they are less
toxic than viral systems, and are not as low-efficient as non-viral systems.
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 465

Author details
Erdal Cevher * and Emre Şefik Çağlar
Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, Beyazıt,
Istanbul, Turkey

Ali Demir Sezer


Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Marmara University,
Haydarpaşa, Istanbul, Turkey

4. References
Al-Dosari MS, Gao X. (2009). Nonviral Gene Delivery: Principles, Limitations, and Recent
Progress. American Association of Pharmaceutical Scientist Journal; 11; 671-681.
Altaner C (2008). Prodrug cancer gene therapy. Cancer Letters; 270; 191–201.
Armendáriz-Borunda J, Bastidas-Ramírez BE, Sandoval-Rodríguez A, González-Cuevas J,
Gómez-Meda B, García-Bañuelos J. (2011). Production of first generation adenoviral
vectors for preclinical protocols: Amplification, purification and functional titration.
Journal of Bioscience and Bioengineering; 112; 415–421.
Balazs DA, Godbey WT. (2010). Liposomes for Use in Gene Delivery. Journal of Drug
Delivery; 2011; 1-12.
Bogdansky S. (1990). In Chasin M, Langer R(ed). Natural Polymers as Drug Delivery
Systems. Biodagradable Polymers as Drug Delivery Systems. Marcel Dekker Inc, ABD. pp:
231-261.
Bulut MO, Akar E. (2012). Dendrimerlerin Önemi Ve Kullanım Alanları. Süleyman Demirel
Üniversitesi Teknik Bilimler Dergisi: 1; 5-11.
Bupp K, Roth MJ. (2002). Altering Retroviral Tropism Using a Random-Display Envelope
Library. Molecular Therapy; 5; 329-335.
Campos SK, Barry MA. (2007). Current Advances and Future Challenges in Adenoviral
Vector Biology and Targeting. Current Gene Therapy: 7; 189-204.
Conwell CC, Huang L, In K. Taira, K. Kataoka, T. Niidome (ed). (2005). Recent Progress in
Non-viral Gene Delivery. Non-viral Gene Therapy Gene Design and Delivery. Springer-
Verlag Tokyo. Japan; pp: 3-11.
Coughlan L, Alba R, Parker A, Bradshaw AC, McNeish IA, Nicklin SA Baker AH. (2010).
Tropism-Modification Strategies for Targeted Gene Delivery Using Adenoviral Vectors.
Viruses: 2; 2290-2355.
Dass CR, Choong PFM. (2006). Selective gene delivery for cancer therapy using cationic
liposomes: In vivo proof of applicability. Journal of Controlled Release; 113; 155–163.
Davies JC, Geddes DM, Alton EWFW. (2003). Pulmonary Gene Therapy. Pharmaceutical Gene
Delivery Systems. Eastern Hemisphere Distribution, ABD. Chapter 14.

* Corresponding Author
466 Recent Advances in Novel Drug Carrier Systems

Davis ME. (2002). Non-viral gene delivery systems. Current Opinion in Biotechnology: 13: 128–
131
Dinh AT, Theofanous T, Mitragotri S. (2005). A Model for Intracellular Trafficking of
Adenoviral Vectors. Biophysical Journal; 89; 1574–1588.
Dufes C, Uchegbu IF, Schatzlein AG. (2005). Dendrimers in gene delivery. Advanced Drug
Delivery Reviews; 57; 2177– 2202.
El-Aneed A. (2004). An overview of current delivery systems in cancer gene therapy. Journal
of Controlled Release; 94; 1– 14.
Escors D, Brecpot K. (2010). Lentiviral vectors in gene therapy: their current status and
future potential. Archivum Immunologiae et Therapia Experimentalis;58;107–119.
Etienne-Grimaldi MC, Bennouna J, Formento JL, Douillard JY, Francoual M, Hennebelle I,
Chatelut E, Francois E, Faroux R, El Hannani C, Jacob JH, Milano G. (2012).
Multifactorial pharmacogenetic analysis in colorectal cancer patients receiving 5-
fluorouracil-based therapy together with cetuximab-irinotecan. British Journal of Clinical
Pharmacology; 73; 776-785.
Felgner PL, Gadek TR, Holm M, Roman R, Chan HW, Wenz M, orthrop JP, Ringold
GM, Danielsen M. (1987) Lipofection: a highly efficient, lipid-mediated DNA-
transfection procedure. Proceedings of the National Academy of Sciences of U.S.A; 84; 7413–
7417.
Ferrara KW. (2010). Driving delivery vehicles with ultrasound. Advanced Drug Delivery
Reviews; 60: 1097–1102.
Freed EO. (1997). Retroviruses. Encyclopedia of Cancer:4; 167-172
Frenkel V. (2008). Ultrasound mediated delivery of drugs and genes to solid tumors
Advanced Drug Delivery Reviews; 60: 1193–1208
Gao X, Kim KS, Liu D. (2007). Nonviral Gene Delivery: What We Know and What Is Next.
American Association of Pharmaceutical Scientist Journal; 9: 92-104.
Genç R. (2008). Dendrimerler: Yeni Nesil Polimerik Ağaçlar. Katalizör: 1; 24-28.
Godbey WT, Mikos AG. (2001). Recent progress in gene delivery using non-viral transfer
Complexes. Journal of Controlled Release: 72; 115–125.
Gönen Korkmaz C. (2008). Gen Transferi-Transfeksiyon: Prostat Kanseri STAMP2 Stabil
Hücre Hattının Oluşturulması. DEÜ Tıp Fakültesi Dergisi; 22: 39 – 45.
He CH, Tabata Y, Gao JQ. (2010). Non-viral gene delivery carrier and its three-dimensional
transfection system. International Journal of Pharmaceutics:386; 232–242.
Hofland HEJ, Shephard L, Sullivan SM. (1996). Formation of stable cationic lipid/DNA
complexes for gene transfer. Proceedings of the National Academy of Sciences; 93: 7305-
7309.
Howarth JL, Lee YB, Uney JB, (2010). Using Viral Vectors as Gene Transfer Tools. Cell
Biology Toxicology; 26; 1-20.
Hu WS, Pathak VK (2000). Design of Retroviral Vectors and Helper Cells for Gene Therapy.
Pharmacological Rewievs; 52; 494-507.
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 467

Smith JG, Wiethoff CM, Phoebe LS, Nemerow GR. (2010). Adenovirus. Current Topics in
Microbiology and Immunology: 343; 195-224.
Ji J, Ji SY, Yang JA, He X, Yang XH, Ling WP, Chen XL. (2012). Ultrasound-targeted
transfection of tissue-type plasminogen activator gene carried by albumin nanoparticles
to dog myocardium to prevent thrombosis after heart mechanical valve replacement.
International Journal of Nanomedicine;7;2911-2919.
Karlsson S, Papayannopoulou T, Schweiger S (1987). Retroviral-mediated transfer of
genomic globin genes leads to regulated production of RNA and protein. Proceedings of
the National Academy of Sciences; 84; 2411-2415.
Katz MG, Fargnoli AS, Pritchette LA, Bridges CR. (2012). Gene delivery technologies for
cardiac applications. Gene Therapy;19;656-669.
Kairouz V, Lipskaia L, Hajjar RJ, Chemaly ER. (2012). Molecular targets in heart failure gene
therapy: current controversies and translational perspectives. Annals of the New York
Academy of Sciences; 1254; 42-50.
Khalid S, Bond JB, Holyoake J, Hawtin RW, Sansom MSP (2008). DNA and lipid bilayers:
self-assembly and insertion. Journal of the Royal Society Interface; 5; 241–250.
Khare R, Chen CY, Weaver EA, Barry MA. (2011). Advances and Future Challenges
in Adenoviral Vector Pharmacology and Targeting. Current Gene Therapy: 11;
241-258.
Kim T, Rothmund T, Kissel T, Kim SW. (2011). Bioreducible polymers with cell penetrating
and endosome buffering functionality for gene delivery systems. Journal of Controlled
Release: 152; 110–119.
Kitamura T, Koshino Y, Shibata F, Oki T, Nakajima H, Nosaka T, Kumagai H. (2003).
Retrovirus-mediated gene transfer and expression cloning: powerful tools in functional
genomics. Experimental Hematology; 31;1007-1014.
Klink D, Schindelhauer D, Laner A, Tucker T, Bebok Z, Schwiebert EM, Boyd AC, Scholte
BJ. (2004). Gene delivery systems—gene therapy vectors for cystic fibrosis. Journal of
Cystic Fibrosis; 3; 203– 212.
Kozarsky KF (2001). Gene Therapy for Cardiovascular Disease. Current Opinion in
Pharmacology 2001; 1; 197–202.
Lagisetty KH, Morgan RA. (2012). Cancer therapy with genetically-modified T cells for the
treatment of melanoma. Journal of Gene Medicine; 14; 400-404.
Lin MTS, Pulkininen L, Uitto J, Yoon K. (2000). The Gene Gun: Current Applicationsin
Cutaneous GeneTherapy. International Journal of Dermatology; 39; 161-170.
Logue JS, Morrison DK. (2012). Complexity in the signaling network: insights from the use
of targeted inhibitors in cancer therapy. Genes & Development; 26; 641-650.
Luo Y, Kofod-Olsen E, Christensen R, Sørensen CB, Bolund L. (2012). Targeted genome
editing by recombinant adeno-associated virus (rAAV) vectors for generating
genetically modified pigs. Journal of Genetics and Genomics; 39; 269-274.
Majhen D, Ambriovic-Ristov A. (2006). Adenoviral vectors-How to use them in cancer gene
therapy? Virus Research;119; 121–133.
468 Recent Advances in Novel Drug Carrier Systems

Mátrai J, Chuah MKL, Vanden Driessche T. (2009). Recent Advances in Lentiviral Vector
Development and Applications. Molecular Therapy; 18: 477–490.
McDonald PC, Winum JY, Supuran CT, Dedhar S. (2012). Recent developments in targeting
carbonic anhydrase IX for cancer therapeutics. Oncotarget; 3, 84-97.
Medina-Kauwe LK. (2003). Endocytosis of adenovirus and adenovirus capsid proteins.
Advanced Drug Delivery Reviews; 55;1485-1496.
Mhashilkar A, Chada S, Roth JA, Ramesh R. (2001). Gene therapy; Therapeutic approaches
and implications. Biotechnology Advances; 19; 279-297.
Miyazaki M, Obata Y, Abe K, Furusu A, Koji T, Tabata Y,Kohno S. (2006). Technological
Advances in Peritoneal Dialysis Research: Gene Transfer Using Nonviral Delivery
Systems. Peritoneal Dialysis International; 26; 633–640.
Miyazaki Y, Miyake A, Nomaguchi A, Adachi A. (2011). Structural dynamics of retroviral
genome and the packaging. Frontiers in Microbiology; 2; 2-9.
Mortimer I, Tam P, MacLachlan I, Graham RW, Saravolac EG, Joshi PB. (1999). Cationic
lipid-mediated transfection of cells in culture requires mitotic activity. Gene Therapy; 6;
403–411.
Mrsny R. (2005). In Amiji MM (ed). Tissue-and Cell-Specific Targeting for the Delivery of
Genetic Information. Polimeric Gene Delivery. CRC PRESS, USA. pp: 1-30.
Muzzonigro TS, Ghivizzani SC, Robbins PD, Evans CH. 1999. The Role of Gene Therapy:
Fact or Fiction? Clinics in Sports Medicine; 18; 223-237.
Navarro J, Risco, Toschi M,Schattman G. (2008). Gene Therapy and Intracytoplasmatic
Sperm Injection (ICSI) – A Review. Placenta; 29; S193–S199.
Niidome T, Huang L. (2002). Gene Therapy Progress and Prospects: Nonviral vectors. Gene
Therapy; 9; 1647–1652.
Olsen JC, Swanstorm R. (1985). A New Pathway in the Generation of Defective Retrovirus
DNA. Journal of Virology; 56; 779-789.
Osten P, Grinevich V, Cetin A. (2007). Viral Vectors: A Wide Range of Choices and High
Level Services. HEP; 178; 177-202.
Pages JC, Danos O. (2003). In Rolland A, Sullivan SM. Retrovectors Go Forward.
Pharmaceutical Gene Delivery Systems. Eastern Hemisphere Distribution, USA. Chapter
9.
Pillai O, Panchagnula R. (2001). Polymers in drug delivery. Current Opinion in Chemical
Biology; 5: 447–451.
Pitt WG, Husseini GA, Staples BJ. (2004). Ultrasonic Drug Delivery – A General Review.
Expert Opinion on Drug Delivery; 1; 37–56.
Prokop A, Davidson JM. (2007). In Lanza R, Langer R, Vacanti J (ed). Gene Delivery into
Cells and Tissues. Princeples of Tissue Engineering. Elsevier Academic Press, ABD; pp:
493-515.
Restifo NP, Dudley ME, Rosenberg SA. (2012). Adoptive immunotherapy for cancer:
harnessing the T cell response. Nature Reviews Immunology; 12; 269-281.
Gene Delivery Systems: Recent Progress in Viral and Non-Viral Therapy 469

Reynolds PN, Feng M, Curiel DT. (1999). Chimeric viral vectors – the best of both worlds?
Molecular Medicine Today; 4; 25-31.
Robbins PD, Ghivizzani SC. (1998). Viral Vectors for Gene Therapy. Pharmacology &
Therapeutics; 80; 35–47.
Schatzlein AG. (2003). Targeting of Synthetic Gene Delivery Systems. Journal of Biomedicine
and Biotechnology: 2; 149–158.
Scheier B, Amaria R, Lewis K, Gonzalez R. (2011). Novel therapies in melanoma.
Immunotherapy; 3;1461-1469.
Somiari S, Glasspool-Malone J, Drabick JJ, Gilbert RA, Heller R, Jaroszeski MJ, Malone RW.
(2000). Theory and in vivo Application of Electroporative Gene Delivery. Molecular
Therapy: 2; 178-187.
Suda T, Liu D. (2007). Hydrodynamic Gene Delivery: Its Principles and Applications.
Molecular Therapy; 15: 2063–2069.
Sullivan SM. (2003). In Sullivan SM, Rolland A (ed). Introduction to Gene Therapy and
Guidelines to Pharmaceutical Development. Pharmaceutical Gene Delivery Systems.
Eastern Hemisphere Distribution, USA; pp. 17-31.
Tunçay M, Çalış S. (1999) İlaç taşıyıcı sistemlerde kullanılan biyoparçalanabilir sentetik ve
doğal polimerler. FABAD J. Pharm. Sci; 24: 109-123.
Uyechi-O’Brien LS, Szoka FC (2003). In Rolland A, Sullivan SM (ed). Mechanisms for
Cationic Lipids in Gene Transfer. Pharmaceutical Gene Delivery Systems. Eastern
Hemisphere Distribution, USA. Chapter 4.
Wang X, Cao L, Wang Y, Wang X, Liu N, You Y. (2012). Regulation of let-7 and its target
oncogenes (Review). Oncology Letters; 3; 955-960.
Ward BB, Baker Jr. JR. (2008). In Majoros IJ, Baker Jr. JR (ed). Targeted Drug Delivery in
General, New Technology in Medicine. Dendrimer Based Nanomedicine. Pan Stanford
Publishing, USA. pp: 1-16.
Willemejane J, Mir LM. (2009). Physical Methods of Nucleic Acid Transfer: General
Concepts and Applications. British Journal of Pharmacology; 157; 207-219.
Witlox MA, Lamfers ML, Wuisman PI, Curiel DT, Siegal GP. (2007). Evolving gene therapy
approaches for osteosarcoma using viral vectors: review. Bone; 40; 797-812.
Wong SY, Pelet JM, Putnam D. (2007). Polymer systems for gene delivery-Past, present, and
future. Progress in Polymer Science; 32; 799–837.
Wunderbaldinger P, Bogdanov Jr A, Weissleder R. (2000). New approaches for imaging in
gene therapy. European Journal of Radiology; 34; 156–165.
Yaron Y, Kramer RL, Johnson MP, Evans MI. (1997). Gene therapy: Is the Future Here Yet?
Fetal Diagnosis and Therapy; 24; 179-197.
Yi Y, Noh MJ, Lee KH. (2011). Current Advances in Retroviral Gene Therapy. Current Gene
Therapy: 11; 218-228.
Zhang X, Edwards JP, Mosser DM. (2009). The Expression of Exogenous Genes in
Macrophages: Obstacles and Opportunities. Methods in Molecular Biology; 531; 1-16.
470 Recent Advances in Novel Drug Carrier Systems

Ziello JE, Huang Y, Jovin IS. (2010). Celular Endocytosis and Gene Delivery. Molecular
Medicine; 16; 222-229.

You might also like